WO1990003791A1 - Methods for interfering with hiv multiplication and composition for the treatment of aids - Google Patents

Methods for interfering with hiv multiplication and composition for the treatment of aids Download PDF

Info

Publication number
WO1990003791A1
WO1990003791A1 PCT/US1989/004249 US8904249W WO9003791A1 WO 1990003791 A1 WO1990003791 A1 WO 1990003791A1 US 8904249 W US8904249 W US 8904249W WO 9003791 A1 WO9003791 A1 WO 9003791A1
Authority
WO
WIPO (PCT)
Prior art keywords
heparin
hiv
molecular weight
cells
tion
Prior art date
Application number
PCT/US1989/004249
Other languages
French (fr)
Inventor
Nicola Diferrante
Original Assignee
Nicola Diferrante
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nicola Diferrante filed Critical Nicola Diferrante
Publication of WO1990003791A1 publication Critical patent/WO1990003791A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H11/00Compounds containing saccharide radicals esterified by inorganic acids; Metal salts thereof

Definitions

  • This invention relates to antiviral agents which inter ⁇ fere with viral intracellular multiplication.
  • a mutated virus may not be recognized by antibodies elicited by a slightly different antigen derived from a previous mutation.
  • T-4 viral envelope glycoproteins which engage with the cell receptors prior to internalization of the whole virus
  • CD4 antigen soluble fragments of T-4 cell viral receptors
  • interference should be a permanent and complete one if it were to prevent healthy cells from becom ⁇ ing infected either from endogenous virus released by dying cells or exogenous virus acquired through the usual means of transmission.
  • An inexpensive, easily available, low molecu ⁇ lar weight, non-protein compound, capable of diffusing throughout the organism and capable of binding the virus or the viral receptors would be more suitable to this purpose than bulky peptides or proteins of finite half-life, costly preparation and potential immunogenic properties.
  • the present invention provides low molecular wight degra ⁇ dation products of naturally occurring linear polyanion heparin (glycosaminoglycan, acid mucopolysaccharide) lacking antithrombic and anticoagulant activities (i.e., less than 50 units antithro bin activity) , yet retaining sufficient nega ⁇ tive charges, as effective agents which interfere with the replication of the human immunodeficiency virus (HIV) .
  • the methods of invention provide for inhibiting T cell death and replication of human immunodeficiency virus (HIV) comprising exposing the human T cells to a pharmaceutically effective amount of a low molecular weight heparin degradation products lacking anticoagulant activity.
  • the present invention fur ⁇ ther provides a method of treating HIV infection by adminis ⁇ tering to the infected host a therapeutically effective amount of a low molecular weight heparin degradation product lacking anticoagulant activity.
  • the administration can be accomplished through any of many routes including intrave ⁇ nously, intraperitoneally, orally, subcutaneously, intramus ⁇ cularly, rectally, topically or by inhalation.
  • heparin is prepared from beef lung or, prefer ⁇ entially from bovine or porcine intestinal mucosa by extrac ⁇ tion procedures based on the high negative charge of mole ⁇ cule. After digestion of the crude material with proteases, the clear supernatant obtained by centrifugation is treated with quaternary ammonium salts. This precipitation may be achieved step-wise, at decreasing salt concentrations, the most highly charged molecules being obtained as a precipitate at the high salt concentration.
  • This higher charged heparin may be precipitated with quaternary ammonium salts above 0.35 M NaCl, while the other contaminating glycosaminglycans (chondroitin sulfates, heparan sulfate) remain in solution, to be precipitated upon further dilution of the solution.
  • the heparin-quaternary ammonium salt precipitate is repeatedly washed in the centrifuge with 95% ethanol/10% potassium acetate in order to remove the quaternary ammonium salt.
  • the potassium salt of heparin is dissolved in water, cleared by centrifugation and further purified by column chromatography on anion or cation exchange resins.
  • hepa ⁇ rin fractions of different molecular weight Several methods are available for the isolation of hepa ⁇ rin fractions of different molecular weight: a) anion ex ⁇ change chromatography in which the heparin is retained on the column at low ionic strength and fractions of increasing molecular weight may be eluted by step-wise increase in the ionic strengths of the eluting solution (from 0.2 M to 3.0 M NaCl) ; or b) gel filtration, in which larger fractions emerge first from the column, to be followed by gradually smaller ones. In this technique, the influence of sulfate content may be minimized by using as eluant a solution of 0.2 M NaCl.
  • Heparin and heparan sulfate are glycosminoglycans which uniquely contain N-sulfate groups. Older methods of de-N- sulfation with controlled hydrolysis in dilute acids (0.04N HC1 at 56° for 4 hours) caused hydrolysis of N-sulfated groups accompanied by some cleavage of glycosidic linkages (depolymerization) and of 0-sulfate groups (de-O-sulfation) .
  • the pyridinium salt of N-sulfated glucosamine residues (present in heparin and heparan sulfate) is desulfated much more rapidly and selec ⁇ tively in dimethyl sulfoxide containing small amounts of water or methanol.
  • the sodium salt or heparin (molecular weight 12,00 to 15,000) is dissolved in 25 ml of water and the solution is passed through a column of Dowex 50x8 at 4° C. The effluent and washing are combined, neutralized with pyridine and lyophilized to give the pyridinium salt of heparin.
  • This salt is dissolved in 100 ml of dimethyl sulfoxide containing 5% water and the solution is kept for 90 minutes at 50 * C, then diluted with equal volume of water.
  • the pH of the solution is adjusted to 9.0-9.5 with 0.1 M NaOH, dialyzed over night against tap water and then against distilled water for 20 hours. Filtration and lyophilization of the retentate gives the sodium salt of completely de-N-sulfated heparin.
  • This large molecular weight compound (12,000 to 15,000 daltons) has no anticoagulant activity.
  • This compound may be acetylated by the method of Danishefsky, et al. Arch. Biochem. , Biophys. 90:114 (1960).
  • Antithro bin (commercially available) bound to an insoluble matrix (Sepharose) may be used to separate large MW heparin into two fractions: one with high and one with low affinity for antithrombin (HA and LA respectively) .
  • the first has approximately 300 U anticoagulant activity/mg; the second is essentially inactive. This technique has allowed to identify the structure of the pentasaccharide of heparin responsible for binding to antithrombin.
  • the internal glucosamine unit carries a 0-sulfate group in position C-3, previously consid ⁇ ered essential for interaction with antithrombin and absent in LA heparin. More recently, however, the consensus is that the 3-O-sulfate is an artifact produced during deamination of the 2-N-sulfate group, which migrates to position 3. In fact, the 3-O-sulfate group is never found in products of bacterial degradation of heparin. However, three additional sulfate groups, are required for high affinity to antithrom ⁇ bin. One of those, 2-N sulfate on the internal glucosamine, may be removed by solvolytic cleavage with methyl sulfoxide, leaving a highly sulfated, nonanticoagulant product.
  • Both these fractions may be depolymerized with nitrous acid, to obtain fragments of various length which may be separated by gel chromatography. It is well documented that anticoagulant activity of heparin is highly dependent on molecular weight. Thus, oligosaccharides of less than 18 monosaccharides (less than 5,000 MW) length have lost their anticoagulant activity but retained their anti ⁇ thrombic activity (anti-Xa activity) .
  • N-sulfated glucosamine is transformed into 2,5- anhydromannose.
  • This residue located at the reducing end of the oligosaccharide products, may be conveniently reduced with sodium borohydride labeled with tritium (NaB 3 H 4 ) , to yield tritium-labeled mannose at the reducing end.
  • This labeling technique allows one to follow the fate of these oligosaccharides 1 , once added to a cell culture.
  • heparinase II ob ⁇ tained originally from Flavobacterium heparinum and now commercially available.
  • 200 mg of heparin in 0.005 M phos ⁇ phate buffer pH 7.0, containing 0.02% bovine serum albumin is incubated at 25° C for up to 125 hours with heparinase II (10 units in 10 ml buffer) .
  • heparinase II 10 units in 10 ml buffer
  • a series of disaccharides is obtained, which may be separated by high pressure liquid chromatography. Shorter periods of incubation, with the enzyme, may yield trisaccharides or oligosaccharides which may be separated by gel chromatography. All these products
  • Sample B used in these experiments is a degradation product of heparin (MW 4.000-5,000 daltons) prepared by mild oxidation of heparin with copper, H 2 02 and ascorbate.
  • the desired sample was then desalted, by gel filtration and precipitated with methanol and dried with ether as de ⁇ scribed in 3.
  • the virus a source of reverse transcriptase
  • concentrations 1.0; 0.3; 0.1.
  • Concentra ⁇ tion 1 is arbitrary and it is defined, for the purpose of this assay, as that amount of virus which provides sufficient enzyme to cause incorporation of tritiated thymidine triphos- phate (3 H TTP) into the template to the extent of 40,000 cpm.
  • the two heparin preparations A and B have been used at sever ⁇ al, decreasing concentrations: 1.0; 0.3; 0.1; 0.03 mg/ml.
  • HIV virus suspension HAV pp213, stock of UTMB Galveston
  • the concentration of the virus (1:10 dilution from the stock solution) was adopted so as not to kill the cells immediately but nevertheless to provide total infection of the cells within 4-5 days.
  • cells were exposed to the heparin preparations A or B by adding the heparin sample to the cells 1 hour before or 1 hour after challenging with virus.
  • CPE cytopathic effects
  • Control cells + Heparin B * for each of # 1.0; 0.3; 01.; and 0.03mg/ml
  • Heparin B protects the cells from being killed by the virus even though they had been infected for 48 hours. Control cells, infected but not treated show morphological changes and fluorescence by day 2 and 3, with cell death by day 5.
  • Control cells plus Heparin B do not show changes of toxicity. However, cells infected at time 0 and treated 48 hours later do not show cytopathic effects, even though they display the presence of viral core protein (P-24) .
  • Heparin B administered 48 hours after viral infection does not prevent the synthesis of the core P-24 antigen of the virus. The latter apparently must have had time to shed the glycoprotein coat, to free its RNA from the core protein, to have the RNA transcripted into DNA and this incorporated into host DNA.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Heparin and low molecular weight derivatives of heparin lacking anticoagulant activity are effective in interfering with the life cycle of HIV. Cells exposed to heparin or its low molecular weight derivatives one hour before or after challenge with HIV showed no evidence of viral infection when examined four to six days post HIV challenge. Control cells similarly challenged with HIV showed 100 % HIV infection six days post HIV challenge. Cells challenged with HIV as much as 48 hours before exposure to heparin or its low molecular derivatives showed little evidence of HIV multiplication and evidenced healthy morphological features.

Description

METHODS FOR INTERFERING WITH HIV MULTIPLICATION AND COMPOSITION FOR THE TREATMENT OF AIDS
SPECIFICATION
FIELD OF THE INVENTION
This invention relates to antiviral agents which inter¬ fere with viral intracellular multiplication.
BACKGROUND OF THE INVENTION
The progressive spreading of the AIDS infection with its tragic consequences on the world population has mobilized worldwide scientific efforts to contain the epidemic and search for effective means to alter the HIV virus life cycle.
The experience acquired in the past with other human viral diseases has immediately suggested that vaccines against the virus be considered and implemented. To this extent, either biosynthetic or extractive fragments of viral glycoprotein envelope, incorporated into appropriate biolog¬ ical carriers, have been injected into animals to assess whether they would be immunogenic (that is capable of causing production of antibodies) and whether the antibodies produced would interact with the virus and prevent its binding to the cells and receptors. Hopefully such an interaction would ameliorate the conditions of the affected subjects and would protect from infection those newly exposed to the virus. As of now, these efforts have failed with respect to the HIV virus since the antibodies, when produced, have not been effective in arresting the viral spread nor the progression of the disease. This ineffectiveness may be caused, at least in part, by the high rate of mutation of the virus, with corresponding changes in the structure of all its gene prod¬ ucts, including the envelope glycoproteins. Thus, a mutated virus may not be recognized by antibodies elicited by a slightly different antigen derived from a previous mutation.
The same arguments may apply to antibodies against the viral reverse transcriptase. This enzyme is responsible for the transcription of the viral genome - RNA - into the (cor¬ responding) DNA which may initiate progeny formation or be incorporated in the genome of the host cell. Moreover, the difficulty of these antibodies to reach the intracellular compartments where the reverse transcriptase is active make their potential usefulness rather questionable.
Other approaches contemplate (1) the synthesis of those peptide fragments of the T-4 viral envelope glycoproteins which engage with the cell receptors prior to internalization of the whole virus; (2) the production of antibodies to the cell viral receptors; and (3) the biosynthesis of soluble fragments of T-4 cell viral receptors (CD4 antigen) capable of intercepting free, extracellular virus and binding it to prevent subsequent internalization. The first two products, by binding and occupying the viral receptors of the sensitive cells would make them unavailable for binding the virus. Considering the variety and the number of cells sensitive to viral attack present in a given organism, and the number of their viral receptors, one wonders whether those peptides or antibodies could be made available with a half-life and in amounts sufficient to keep the viral receptors constantly occupied for a finite amount of time. Yet, approaches to interfere with the binding of the virus to the surface receptors of the sensitive cells are sound because, if successful, they would prevent the inter¬ nalization of the virus which would remain exposed to the other humoral and cellular mechanisms of defense of the organism and would be prevented from incorporating its genome into that of the host cell.
However, such interference should be a permanent and complete one if it were to prevent healthy cells from becom¬ ing infected either from endogenous virus released by dying cells or exogenous virus acquired through the usual means of transmission. An inexpensive, easily available, low molecu¬ lar weight, non-protein compound, capable of diffusing throughout the organism and capable of binding the virus or the viral receptors would be more suitable to this purpose than bulky peptides or proteins of finite half-life, costly preparation and potential immunogenic properties.
SUMMARY OF THE INVENTION
The present invention provides low molecular wight degra¬ dation products of naturally occurring linear polyanion heparin (glycosaminoglycan, acid mucopolysaccharide) lacking antithrombic and anticoagulant activities (i.e., less than 50 units antithro bin activity) , yet retaining sufficient nega¬ tive charges, as effective agents which interfere with the replication of the human immunodeficiency virus (HIV) . The methods of invention provide for inhibiting T cell death and replication of human immunodeficiency virus (HIV) comprising exposing the human T cells to a pharmaceutically effective amount of a low molecular weight heparin degradation products lacking anticoagulant activity. The present invention fur¬ ther provides a method of treating HIV infection by adminis¬ tering to the infected host a therapeutically effective amount of a low molecular weight heparin degradation product lacking anticoagulant activity. The administration can be accomplished through any of many routes including intrave¬ nously, intraperitoneally, orally, subcutaneously, intramus¬ cularly, rectally, topically or by inhalation.
DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS
Heparin and Its Degradation Products.
Commercial heparin is prepared from beef lung or, prefer¬ entially from bovine or porcine intestinal mucosa by extrac¬ tion procedures based on the high negative charge of mole¬ cule. After digestion of the crude material with proteases, the clear supernatant obtained by centrifugation is treated with quaternary ammonium salts. This precipitation may be achieved step-wise, at decreasing salt concentrations, the most highly charged molecules being obtained as a precipitate at the high salt concentration.
This higher charged heparin may be precipitated with quaternary ammonium salts above 0.35 M NaCl, while the other contaminating glycosaminglycans (chondroitin sulfates, heparan sulfate) remain in solution, to be precipitated upon further dilution of the solution.
The heparin-quaternary ammonium salt precipitate is repeatedly washed in the centrifuge with 95% ethanol/10% potassium acetate in order to remove the quaternary ammonium salt. The potassium salt of heparin is dissolved in water, cleared by centrifugation and further purified by column chromatography on anion or cation exchange resins.
Several methods are available for the isolation of hepa¬ rin fractions of different molecular weight: a) anion ex¬ change chromatography in which the heparin is retained on the column at low ionic strength and fractions of increasing molecular weight may be eluted by step-wise increase in the ionic strengths of the eluting solution (from 0.2 M to 3.0 M NaCl) ; or b) gel filtration, in which larger fractions emerge first from the column, to be followed by gradually smaller ones. In this technique, the influence of sulfate content may be minimized by using as eluant a solution of 0.2 M NaCl.
The relevance of these techniques to the present inven¬ tion depends on the linear correlation between molecular size and anticoagulant activity of heparin. We are particularly interested in heparin fractions lacking anticoagulant activi¬ ty, yet still capable of interfering with the multiplication of the virus and still possessing the properties of stimulat¬ ing cellular endogenous mechanisms of defense. Other method¬ ologies that may be used for the same purpose include affini¬ ty chromatography of heparin on antithrombin-sepharose col¬ umns and factor Xa-sepharose columns in order to separate the high affinity fraction, having very high antithrombic or anticoagulant activity, from the low - affinity ones, essen¬ tially devoid of such activities. Thus, with these tech¬ niques, we were able to screen large and small molecular weight fractions of heparin without dangerous anticoagulant activity, for any possible inhibitory activity on the multi¬ plication of the virus.
Additional chemical and enzymic methods may be used to degrade large molecular weight heparin fractions without further affecting their sulfate content. Dietrick, C.P., Biochem. J. 108: 647-654 (1968). Low molecular weight heparin (from porcine intestinal mucosa) is produced by ni¬ trous acid degradation of native heparin, to a 4,000-6,000 MW. It is obtainable commercially through Sigma Company. This compound still possesses good anti-Xa activity, (> 150 IU/mg) but lower antithrombic activity (>40 IU/mg) than the undegraded original material. This low molecular weight heparin can be completely de-N-sulfated and N-acetylated as described below, in order to obtain a product without anti¬ coagulant activity. Heparin and heparan sulfate are glycosminoglycans which uniquely contain N-sulfate groups. Older methods of de-N- sulfation with controlled hydrolysis in dilute acids (0.04N HC1 at 56° for 4 hours) caused hydrolysis of N-sulfated groups accompanied by some cleavage of glycosidic linkages (depolymerization) and of 0-sulfate groups (de-O-sulfation) . More recently, it has been reported that the pyridinium salt of N-sulfated glucosamine residues (present in heparin and heparan sulfate) is desulfated much more rapidly and selec¬ tively in dimethyl sulfoxide containing small amounts of water or methanol. The sodium salt or heparin (molecular weight 12,00 to 15,000) is dissolved in 25 ml of water and the solution is passed through a column of Dowex 50x8 at 4° C. The effluent and washing are combined, neutralized with pyridine and lyophilized to give the pyridinium salt of heparin.
This salt is dissolved in 100 ml of dimethyl sulfoxide containing 5% water and the solution is kept for 90 minutes at 50* C, then diluted with equal volume of water. The pH of the solution is adjusted to 9.0-9.5 with 0.1 M NaOH, dialyzed over night against tap water and then against distilled water for 20 hours. Filtration and lyophilization of the retentate gives the sodium salt of completely de-N-sulfated heparin. This large molecular weight compound (12,000 to 15,000 daltons) has no anticoagulant activity. This compound may be acetylated by the method of Danishefsky, et al. Arch. Biochem. , Biophys. 90:114 (1960). 300 micrograms of the sodium salt of completely de-N-sulfated heparin in 30 ml of water are added to 5 ml of methanol and 0.9 ml of acetic anhydride. The mixture is kept for two hours at pH 6.5 by adding 10% NaHC03, under stirring at 3-4° C. The reaction mixture is dialyzed against running tap water over night and then against distilled water for 20 hours. Filtration and lyophilization of the retentate gives the sodium salt of completely de-N-sulfated, N-acetylated heparin. If 14C, 3H or deuterium-labeled acetic anhydride is used, the final acety- lated product will be conveniently labeled. Such a radioac- tively labeled derivative may be useful in following the fate of large molecular weight heparin added to a cell culture or administered to experimental animals.
Another facile and fruitful approach to the preparation of non anticoagulant derivatives of heparin is provided by E. Holmes et al., Hemostasis 16, Supplement 2: 1-7 (1986). Antithro bin (commercially available) bound to an insoluble matrix (Sepharose) may be used to separate large MW heparin into two fractions: one with high and one with low affinity for antithrombin (HA and LA respectively) . The first has approximately 300 U anticoagulant activity/mg; the second is essentially inactive. This technique has allowed to identify the structure of the pentasaccharide of heparin responsible for binding to antithrombin. The internal glucosamine unit carries a 0-sulfate group in position C-3, previously consid¬ ered essential for interaction with antithrombin and absent in LA heparin. More recently, however, the consensus is that the 3-O-sulfate is an artifact produced during deamination of the 2-N-sulfate group, which migrates to position 3. In fact, the 3-O-sulfate group is never found in products of bacterial degradation of heparin. However, three additional sulfate groups, are required for high affinity to antithrom¬ bin. One of those, 2-N sulfate on the internal glucosamine, may be removed by solvolytic cleavage with methyl sulfoxide, leaving a highly sulfated, nonanticoagulant product.
Both these fractions (HA and LA) may be depolymerized with nitrous acid, to obtain fragments of various length which may be separated by gel chromatography. It is well documented that anticoagulant activity of heparin is highly dependent on molecular weight. Thus, oligosaccharides of less than 18 monosaccharides (less than 5,000 MW) length have lost their anticoagulant activity but retained their anti¬ thrombic activity (anti-Xa activity) .
Following the procedures of Pejlaer, G. et al., J. Biol. Chem. 263:5197 (1988), 42 micrograms of sodium nitrate (NaN02) are added to one microgram of heparin in 20 ml of ice-cold H20, acidified to pH 1.4 with dilute H2S04. After three hours in the ice bath, the oligosaccharides are pre¬ cipitated with 10 volumes ethanol and separated by gel chro¬ matography on sephadex G-50. Fractions composed of 2 to 20 monosaccharides may thus be obtained. The nitrous acid treatment causes an oxidative deamination of N-sulfated glucosamine, with cleavage of the glucosaminidic linkage. Thus, N-sulfated glucosamine is transformed into 2,5- anhydromannose. This residue, located at the reducing end of the oligosaccharide products, may be conveniently reduced with sodium borohydride labeled with tritium (NaB3H4) , to yield tritium-labeled mannose at the reducing end. This labeling technique allows one to follow the fate of these oligosaccharides1, once added to a cell culture.
Finally, another method for degrading heparin to di and trisaccharides makes use of the enzyme heparinase II, ob¬ tained originally from Flavobacterium heparinum and now commercially available. 200 mg of heparin in 0.005 M phos¬ phate buffer pH 7.0, containing 0.02% bovine serum albumin, is incubated at 25° C for up to 125 hours with heparinase II (10 units in 10 ml buffer) . A series of disaccharides is obtained, which may be separated by high pressure liquid chromatography. Shorter periods of incubation, with the enzyme, may yield trisaccharides or oligosaccharides which may be separated by gel chromatography. All these products
1 A note of caution: oligosaccharides prepared by nitrous acid oxidation have been used to produce monoclonal antibodies in mice. Thus, the possible antigenicity of 2,5 anhydro annose must be kept in mind. Pejlaer et al. J. Biol. Chem. 263:5197 (1988) . of enzymic cleavage have a 4-5 unsaturated uronic acid at the non-reducing end and have a MW of about 1,000 to about 3,000 daltons.
Heparin Sample B
Sample B used in these experiments is a degradation product of heparin (MW 4.000-5,000 daltons) prepared by mild oxidation of heparin with copper, H202 and ascorbate.
100 g of sodium heparin were dissolved in 10 liters of a solution containing 0.5M NaCl; 0.35M sodium acetate; 0.35% ascorbic acid; 0.45% copper acetate and H202 to a final concentration of 1.8 volumes. The pH of the solution was adjusted to 7.80 before the addition of peroxide and again immediately after the addition of peroxide.
The solution was incubated at 50* C, with stirring for 20 hours, samples of 5-7 ml were collected at regular inter¬ vals. The buffered solution, prior to addition of peroxide and final pH adjustment, was pale blue-green in color. Upon addition of peroxide and alkali to pH 7.80, vigorous bubbling ensued, with a raise in the solution temperature to 65° C. The bubbling lasted for 30 minutes after which the tempera¬ ture of the solution slowly returned to 50° C.
At the end of the incubation, the solution was clear and blue-green in color. The samples collected at various times during incubation and a final sample were processed in paral¬ lel throughout the following steps:
1. Passage through 7x1 cm columns of cation ex¬ change resin Dowex 50x4, 200 mesh in the hydrogen form in order to retain the copper and allow recovery of free heparin in the effluent. Its elution was followed simply by monitor¬ ing the strong acidity of the effluent. Whenever the latter became neutral, the elution of heparin was complete. 2. Immediate neutralization of each heparin ali¬ quot with sodium hydroxide, in order to obtain the sodium salt of the glycan.
3. The various aliquots were reduced in volume (if needed) , with a rotary evaporator, under vacuum, and/or directly precipitated in the centrifuge with 2 volumes of absolute methanol. After washing twice with absolute metha¬ nol, each precipitate was washed and dried with ether to the powder state. The material obtained at the end of the incu¬ bation was weighed in order to calculate the yield of the process (80-85%) .
4. Each sample collected during and at the end of the incubation was analyzed by electrophoresis on 6% acryla- mide gels, to verify whether degradation had occurred. Sam¬ ples of original and degraded heparin or Fragmin (a trade- marked product of Kabi Vitrum) and of known products of heparin degradation were run as standards. The electropho¬ resis was run with 0.6M barbital buffer pH 8.60 for a period of one hour. Thereafter, the gels were stained with 0.1% Toluidine Blue in 1% acetic acid in water for 30 minutes. Destaining was carried out in 1% acetic acid, with mild agitation. Destained and dried gels were photographed or scanned to obtain a graphic representation of the degree of degradation.
5. When a final sample showed signs of great heterogeneity, with bands or peak indicating degradation products of different sizes, a water solution of the sample was passed through an appropriate size column of anion ex¬ change resin Dowex 1x2, 100-200 mesh, in the chloride form in order to retain the heparin. This was eluted in discreet fraction of increasing molecular size using increasing con¬ centration of NaCl: from 1.0M to 2.0,3.0 and 4.0M NaCl.
Because this techniques eluted the sodium salt of hepa¬ rin, monitoring the effluent acidity was not feasible. Thus, either one has to monitor the absorption of effluent at 310nm or spot a sample of effluent on filter paper impregnated with alcian blue in order to detect the methachromatic reaction of heparin.
The desired sample was then desalted, by gel filtration and precipitated with methanol and dried with ether as de¬ scribed in 3.
While in this series of experiments copper acetate was used as a source of metal catalyst, iron salts, copper or iron turning may be equally used for the production of OH free radicals (Fentόn reaction) .
Experimental
What follows is a description of the assays performed to demonstrate anti reverse transcriptase activity, if any, of
native heparin (Sample A) and Cu"""/H202/ascorbate degraded heparin (Sample B) .
HIV RT ASSAY
lOx Buffer:
1M Tris-HCL, pH 7.4-8.0
1M DTT
1M MgCl2
1M KC1
10% Triton X-100
6mM GSH
Figure imgf000014_0001
100 ul
Template: lOmM Tris + 0.1 mM EDTA 180 ul 0.5mg/ml rAdT 120 ul Add H20 100 ul
400 ul* (Dilute 1:2 Tris:EDTA)
RXN Mixture (1:5) (1:2) lOx Buffer [H3]-TTP:H20 Template Sample*
Figure imgf000014_0002
35.0 ul 50 ul *Supernate of fluid plus cells centrifuged at 4°C.
1. Incubate RXN Mixture 2 hours at 41° C.
2. Spot 20 ul on DE81 strips (handle DE81 paper with gloves on only) .
3. Wash with 5% Na2HP04, 4-6x - 4 min/wash double distilled H20 2x - 1 min/wash
95% EtOH 2x - 1 min/wash
4. Place in scintillation vials and Aquasol. Example I
The virus, a source of reverse transcriptase, was used at three, decreasing concentrations, 1.0; 0.3; 0.1. Concentra¬ tion 1 is arbitrary and it is defined, for the purpose of this assay, as that amount of virus which provides sufficient enzyme to cause incorporation of tritiated thymidine triphos- phate (3HTTP) into the template to the extent of 40,000 cpm. The two heparin preparations A and B have been used at sever¬ al, decreasing concentrations: 1.0; 0.3; 0.1; 0.03 mg/ml.
The data indicate that both preparations cause inhibition of reverse transcriptase, preparation A better than B, with decreasing inhibition at decreasing concentrations. Virus dilution does not seem to enhance inhibitory activity.
Example II
This set of experiments investigated whether the two heparin preparations (A or B) interfere with the multiplica¬ tion of HIV in T cells. Both heparin preparations were added to the cells either 1 hour before the virus was added or 1 hour after the virus was added. The rationale for this is that we did not know whether heparin might gain access to the cell interior by itself or whether it needs the virus to proceed - piggy-back - therein. (See, "Animal Viruses Pro¬ mote the Entry of Polysaccharides with Antiviral Activity into Cells," E. Gonzalez E. L. Carrasco. Biochem. Biophvs. Res. Commun. 146; 1303-1310, 1987) .
In these experiments T cells derived from human adult lymphocytic leukemia (ALL) , line C8166, were used at a concentration of 2 x 105cells in 0.5 ml of medium RPMI1640 (standard medium for such experiments) . This amount of cells was place in each well of a multi-well plate. 0.1 ml of HIV virus suspension (HIV pp213, stock of UTMB Galveston) was added to each well and incubation of the plate was carried out at 37° C for up to 21 days.
The concentration of the virus (1:10 dilution from the stock solution) was adopted so as not to kill the cells immediately but nevertheless to provide total infection of the cells within 4-5 days.
In a first set of experiments, cells were exposed to the heparin preparations A or B by adding the heparin sample to the cells 1 hour before or 1 hour after challenging with virus.
Wells were examined every 2 days and samples from wells were removed to measure cellular presence of a viral p-24 core protein antigen. Cells from these samples were spotted in the microscopic fluorescence slides (Cell Line Associates, Inc. 33 Cargo line, Newfield, NJ, fluorescence slides, 8 wells, 8mm) , and fixed with cold acetone and treated with a fluorescent monoclonal antibody against P24. The fluorescent cells in each sample were counted and expressed as a percent¬ age of all cells, using a fluorescence microscope.
TABLE 1: HEPARIN ADDED 1 HOUR BEFORE VIRUS
Concentrations/ml
HEPARIN A
No Heparin Imcr 0.3mcr O.lmg 0.03mσ
DAY 4 30% pos neg neg neg 1-2% pos
DAY 6 100% pos neg neg neg 1-3% pos
HEPARIN B
DAY 4 30% pos neg neg neg neg
DAY 6 100% pos neg neg neg neg TABLE 2: HEPARIN ADDED 1 HOUR AFTER VIRUS ADDITION
n lmg 0.3mg O.lmg 0.03mg neg neg neg 1-3% pos neg neg neg 1-3% pos
neg neg neg neg
Figure imgf000017_0001
neg neg neg neg
CONTROLS: Cells without virus, all negative.
Cells without virus but with heparin A or B at stated concentrations, to investigate for toxicity, as per visual morphological inspection, revealed that Sample A showed +/- toxicity at lmg concentration only, while Sample B did not show toxicity at any concentration.
EXAMPLE II
An additional experiment was performed with Heparin B, to establish any effect on late infection. The test was designed essentially along the lines described in EXAMPLE II. The same cell line, same HIV virus strain, same fluorescence technique, were used to detect P-24 antigen on cell surface. The differences included: 10 the viral inoculum was carried out with full strength virus concentration, in order to obtain in the controls the full effect of the infection in a short period of time; and 2) the Heparin B was added 48 hours later. The virus killing effect was measured by the tripan blue exclusion method. The rest of the experiment was the same. Wells were examined every day for morphological evi¬ dence of cytopathic effects (CPE) (*) and cell samples were obtained every day for fluorescence microscopy to identify presence of P-24 on cell surface (#) . Heparin B was used at l.Omg; 0.3mg; O.lmg and 0.03mg/ml, as previously noted. Both the cytopathic effects and the amount of fluorescence were scored from 1+ to 4+.
TABLE 3: CPE (*=0) AND FLUORESCENCE (#=0)
Days of 0 1 2 3 4 5 6 7 1 experiment
Control cells, not infected *
# # # # # # # # # # #
Control cells, + Heparin B * for each of #
Figure imgf000018_0001
1.0; 0.3; 01.; and 0.03mg/ml
Control cells infected * 1+ 3+ 3+
# 3+ 3+ 3+ 4+
Cells infected + Heparin B for each of * * * * * * * 1.0;; 0.3; # # 3+ 3+ 3+ 3+ 3+ 0.1; 0.03mg/ml
It is clear that Heparin B protects the cells from being killed by the virus even though they had been infected for 48 hours. Control cells, infected but not treated show morphological changes and fluorescence by day 2 and 3, with cell death by day 5.
Control cells plus Heparin B do not show changes of toxicity. However, cells infected at time 0 and treated 48 hours later do not show cytopathic effects, even though they display the presence of viral core protein (P-24) .
These data indicate that Heparin B, administered 48 hours after viral infection does not prevent the synthesis of the core P-24 antigen of the virus. The latter apparently must have had time to shed the glycoprotein coat, to free its RNA from the core protein, to have the RNA transcripted into DNA and this incorporated into host DNA.
The presence on the cell of P-24 antigen clearly indi¬ cates that the virus has undergone a good part of its intra¬ cellular life cycle. Yet, the treated cells do not show signs of suffering, and survive well. Thus, Heparin B apparently acts quite late in the life cycle of the virus, protecting cells from virus killing.
While the presently preferred embodiments of the inven¬ tion have been set forth for the purposes of disclosure, changes and modifications therein can be made which are within the spirit of the invention as defined by the scope of the following claims.

Claims

WHAT IS CLAIMED IS:
1. A method of inhibiting the multiplication of HIV in human T cells comprising exposing the human T cells to a pharmaceutically effective amount of low molecular weight heparin degradation product lacking anticoagulant activity, said heparin degradation product obtained by oxidation of heparin using free radical OH.
2. The method of Claim 1 wherein the heparin degrada¬ tion product has a molecular weight ranging from about 4,000 to about 6,000 daltons.
3. The method of claim 1 wherein the heparin degrada¬ tion product is prepared by mild oxidation of heparin using H202 and a metal catalyst.
4. The method of claim 1 wherein the pharmaceutically effective amount ranges from about lmg/ml to about 0.03 mg/ml.
5. A method of prolonging the survival time of human T cells challenged with the human immunodeficiency virus (HIV) comprising exposing the human T cells to a pharmaceutically effective amount of low molecular weight heparin degradation product lacking anticoagulant activity, said heparin degrada¬ tion product obtained by oxidation of heparin using free radical OH.
6. The method of claim 5 wherein the heparin degrada¬ tion product has a molecular weight ranging from about 4,000 to about 6,000 daltons.
7. The method of claim 5 wherein the heparin degrada¬ tion product is prepared by mild oxidation of heparin.
8. The method of claim 5 wherein the pharmaceutically effective amount ranges from about lmg/ml to about 0.03mg/ml.
9. A method of treating a host infected with human immunodeficiency virus (HIV) comprising administering to the host a pharmaceutically effective amount of a low molecular weight heparin degradation product lacking anticoagulant activity, said heparin degradation product obtained by oxida¬ tion of heparin using free radical OH.
10. The method of claim 9 wherein the heparin degrada¬ tion product has a molecular weight ranging from about 4,000 to about 6,000 daltons.
11. The method of claim 9 wherein the heparin degrada¬ tion product is prepared by mild oxidation of heparin.
12. The method of claim 9 wherein the pharmaceutically effective amount ranges from about lmg/ml to about 0.03mg/ml.
13. The method of claim 9 wherein the heparin degrada¬ tion product is administered orally, intravenously, or intraperitoneally, subcutaneously, intramuscularly, topical¬ ly, rectally or by inhalation.
14. A pharmaceutical composition for treating human immunodeficiency virus (HIV) infection comprising: a pharmaceutically effective amount of a low molecular weight heparin degradation product lacking anticoagulant activity together with a pharmaceutically acceptable diluent, said heparin degradation product obtained by oxida¬ tion of heparin using free radical OH.
15. The pharmaceutical composition of claim 14 wherein the heparin degradation product has a molecular weight rang¬ ing from about 4,000 to about 6,000 daltons.
16. The pharmaceutical composition of claim 14 wherein the heparin degradation product is prepared by mild oxidation of heparin.
17. The pharmaceutical composition of claim 14 wherein the pharmaceutically effective amount ranges from about lmg/ml to about 0.03mg/ml.
PCT/US1989/004249 1988-10-05 1989-09-27 Methods for interfering with hiv multiplication and composition for the treatment of aids WO1990003791A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US25368688A 1988-10-05 1988-10-05
US253,686 1988-10-05

Publications (1)

Publication Number Publication Date
WO1990003791A1 true WO1990003791A1 (en) 1990-04-19

Family

ID=22961299

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1989/004249 WO1990003791A1 (en) 1988-10-05 1989-09-27 Methods for interfering with hiv multiplication and composition for the treatment of aids

Country Status (2)

Country Link
AU (1) AU4426289A (en)
WO (1) WO1990003791A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992022588A1 (en) * 1991-06-14 1992-12-23 Italfarmaco S.P.A. Acylated heparins as inhibitors of the replication of retroviruses
EP0583360A1 (en) * 1991-05-02 1994-02-23 Yeda Research And Development Co. Ltd. Compositions for the prevention and/or treatment of pathological processes
US5861382A (en) * 1992-05-01 1999-01-19 Yeda Research And Development Co. Ltd. Methods for regulation of active TNF-α
US6399078B1 (en) 1998-06-01 2002-06-04 University Of Maryland Biotechnology Institute Chemokine—glycosaminoglycan complexes and their use in treating or preventing receptor mediated diseases
WO2003078960A2 (en) * 2002-03-11 2003-09-25 Momenta Pharmaceuticals, Inc. Analysis of sulfated polysaccharides
US6750207B1 (en) 1992-05-01 2004-06-15 Yeda Research And Development Co. Ltd. Compositions for the regulation of cytokine activity
US7285536B2 (en) 2001-12-05 2007-10-23 Yeda Research And Development Co., Ltd. Anti-cancer therapeutic compounds
US7790466B1 (en) 2007-01-26 2010-09-07 Momenta Pharmaceuticals, Inc. Evaluating mixtures of low molecular weight heparins by chain profiles or chain mapping
CN101942039A (en) * 2010-09-16 2011-01-12 山东海科化工集团有限公司 Parnaparin production method
US7968082B1 (en) 2007-01-26 2011-06-28 Momenta Pharmaceuticals, Inc. Evaluating mixtures of low molecular weight heparins by NMR
US8435795B2 (en) 2010-01-19 2013-05-07 Momenta Pharmaceuticals, Inc. Evaluating heparin preparations
US9068957B2 (en) 2011-02-21 2015-06-30 Momenta Pharmaceuticals, Inc. Evaluating heparin preparations
US9139876B1 (en) 2007-05-03 2015-09-22 Momenta Pharmacueticals, Inc. Method of analyzing a preparation of a low molecular weight heparin

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0355905B1 (en) * 1988-08-24 1993-01-07 Akzo N.V. Heparin fragments and fractions with anti-hiv action

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1986006729A1 (en) * 1985-05-17 1986-11-20 Opocrin S.P.A. Laboratorio Farmacobiologico Depolymerized hexosaminoglucan sulfates endowed with an antithrombotic, fibrinolytic, antiinflammatory activity, their process of preparation and related pharmaceutical compositions

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1986006729A1 (en) * 1985-05-17 1986-11-20 Opocrin S.P.A. Laboratorio Farmacobiologico Depolymerized hexosaminoglucan sulfates endowed with an antithrombotic, fibrinolytic, antiinflammatory activity, their process of preparation and related pharmaceutical compositions

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0583360A1 (en) * 1991-05-02 1994-02-23 Yeda Research And Development Co. Ltd. Compositions for the prevention and/or treatment of pathological processes
EP0583360A4 (en) * 1991-05-02 1994-04-06 Yeda Research And Development Co. Ltd.
US5474987A (en) * 1991-05-02 1995-12-12 Yeda Research And Development Co. Ltd. Methods of using low molecular weight heparins treatment of pathological processes
US5686431A (en) * 1991-05-02 1997-11-11 Yeda Research And Development Co., Ltd. Methods of using low molecular weight heparins for treatment of pathological processes
US5908837A (en) * 1991-05-02 1999-06-01 Yeda Research And Development Co. Ltd. Methods of using low molecular weight heparins for prevention or treatment of pathological processes
WO1992022588A1 (en) * 1991-06-14 1992-12-23 Italfarmaco S.P.A. Acylated heparins as inhibitors of the replication of retroviruses
US5861382A (en) * 1992-05-01 1999-01-19 Yeda Research And Development Co. Ltd. Methods for regulation of active TNF-α
US6020323A (en) * 1992-05-01 2000-02-01 Yeda Research And Development Co. Ltd. Compositions and methods for regulation of active TNF-α
US7332480B2 (en) 1992-05-01 2008-02-19 Yeda Research And Development Co. Ltd. Compositions for the regulation of cytokine activity
US6750207B1 (en) 1992-05-01 2004-06-15 Yeda Research And Development Co. Ltd. Compositions for the regulation of cytokine activity
US6399078B1 (en) 1998-06-01 2002-06-04 University Of Maryland Biotechnology Institute Chemokine—glycosaminoglycan complexes and their use in treating or preventing receptor mediated diseases
US7285536B2 (en) 2001-12-05 2007-10-23 Yeda Research And Development Co., Ltd. Anti-cancer therapeutic compounds
US7947507B2 (en) 2002-03-11 2011-05-24 Momenta Pharmaceuticals, Inc. Analysis of sulfated polysaccharides
WO2003078960A2 (en) * 2002-03-11 2003-09-25 Momenta Pharmaceuticals, Inc. Analysis of sulfated polysaccharides
US7575886B2 (en) 2002-03-11 2009-08-18 Momenta Pharmaceuticals, Inc. Analysis of sulfated polysaccharides
US8715953B2 (en) 2002-03-11 2014-05-06 Momenta Pharmaceuticals, Inc. Analysis of sulfated polysaccharides
WO2003078960A3 (en) * 2002-03-11 2005-01-27 Momenta Pharmaceuticals Inc Analysis of sulfated polysaccharides
US7968082B1 (en) 2007-01-26 2011-06-28 Momenta Pharmaceuticals, Inc. Evaluating mixtures of low molecular weight heparins by NMR
US8076149B1 (en) 2007-01-26 2011-12-13 Momenta Pharmaceuticals, Inc. Evaluating mixtures of low molecular weight heparins by chain profiles or chain mapping
US8252597B1 (en) 2007-01-26 2012-08-28 Momenta Pharmaceuticals, Inc. Evaluating mixtures of low molecular weight heparins by chain profiles or chain mapping
US8617896B1 (en) 2007-01-26 2013-12-31 Zachary Shriver Evaluating mixtures of low molecular weight heparins by chain profiles or chain mapping
US7790466B1 (en) 2007-01-26 2010-09-07 Momenta Pharmaceuticals, Inc. Evaluating mixtures of low molecular weight heparins by chain profiles or chain mapping
US9139876B1 (en) 2007-05-03 2015-09-22 Momenta Pharmacueticals, Inc. Method of analyzing a preparation of a low molecular weight heparin
US8435795B2 (en) 2010-01-19 2013-05-07 Momenta Pharmaceuticals, Inc. Evaluating heparin preparations
CN101942039A (en) * 2010-09-16 2011-01-12 山东海科化工集团有限公司 Parnaparin production method
US9068957B2 (en) 2011-02-21 2015-06-30 Momenta Pharmaceuticals, Inc. Evaluating heparin preparations

Also Published As

Publication number Publication date
AU4426289A (en) 1990-05-01

Similar Documents

Publication Publication Date Title
Oegema Jr et al. Phosphorylation of chondroitin sulfate in proteoglycans from the swarm rat chondrosarcoma.
Pejler et al. Structure and antithrombin-binding properties of heparin isolated from the clams Anomalocardia brasiliana and Tivela mactroides.
Ferguson et al. Glycosyl-phosphatidylinositol moiety that anchors Trypanosoma brucei variant surface glycoprotein to the membrane
US4184917A (en) Process for producing a structurally modified interferon
WO1990003791A1 (en) Methods for interfering with hiv multiplication and composition for the treatment of aids
US4847338A (en) Low molecular weight heparin fragments as inhibitors of complement activation
Dorfman et al. Synthesis of acid mucopolysaccharides by glial tumor cells in tissue culture
Mahieu et al. Chemical structure of tubular and glomerular basement membranes of human kidney: Isolation, purification, carbohydrate and amino acid composition
HUT77134A (en) Keratan sulfate oligosaccharide fraction and drug containing the same and process for preparing the said oligosaccharides
US3737524A (en) Medicaments derived from nucleic acids,processes for their preparation and their use
Thonar et al. An oligosaccharide component in proteoglycans of articular cartilage
Muramatsu et al. Characterization of glycopeptides isolated from membranes of F9 embryonal carcinoma cells
Antonopoulos et al. Isolation and identification of the glycosaminoglycans from fracture callus
US6534648B1 (en) Isolated algal lipopolysaccharides and use of same to inhibit endotoxin-initiated sepsis
JP4043533B2 (en) Low molecular weight lipopolysaccharide
EP0287076B1 (en) Receptor of the minor rhinovirus receptor group
US5712245A (en) Receptor of the minor human rhinovirus receptor group
US5603932A (en) Receptor of the minor human rhinovirus receptor group
Danishefsky et al. Heparin derivatives prepared by modification of the uronic acid carboxyl groups
Lloyd The metabolism of exogenous N-acetyl-d-glucosamine 6-O [35S]-sulphate in the normal rat
Suda et al. Structural characterization of heparin's binding domain for human platelets
Neiderhiser et al. The purification and properties of the glycoproteins of pig gallbladder bile
Oguri et al. Isolation, characterization, and localization of glycosaminoglycans in rabbit bone marrow
Zhuo et al. Irradiation of the bovine mitochondrial F1-ATPase previously inactivated with 5'-p-fluorosulfonylbenzoyl-8-azido-[3H] adenosine cross-links His-beta 427 to Tyr-beta 345 within the same beta subunit.
JP4019128B2 (en) Anti-herpesvirus

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AT AU BR CH DE DK GB JP LU NL SE

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE FR GB IT LU NL SE

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642